You are on page 1of 10

Pharmacological Research 156 (2020) 104792

Contents lists available at ScienceDirect

Pharmacological Research
journal homepage: www.elsevier.com/locate/yphrs

Review

Implication of HMGB1 signaling pathways in Amyotrophic lateral sclerosis T


(ALS): From molecular mechanisms to pre-clinical results
Yam Nath Paudela,*, Efthalia Angelopouloub, Christina Piperib,**, Iekhsan Othmana,
Mohd. Farooq Shaikha,*
a
Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
b
Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece

ARTICLE INFO ABSTRACT

Keywords: Amyotrophic lateral sclerosis (ALS) is a devastating and rapidly progressing neurodegenerative disorder with no
HMGB1 effective disease-modifying treatment up to date. The underlying molecular mechanisms of ALS are not yet
ALS completely understood. However, the critical role of the innate immune system and neuroinflammation in ALS
RAGE pathogenesis has gained increased attention. High mobility group box 1 (HMGB1) is a typical damage-associated
TLR4
molecular pattern (DAMP) molecule, acting as a pro-inflammatory cytokine mainly through activation of its
Motor neurons
Neuroinflammation
principal receptors, the receptor for advanced glycation end products (RAGE) and toll-like receptor 4 (TLR4)
which are crucial components of the innate immune system. HMGB1 is an endogenous ligand for both RAGE and
TLR4 that mediate its biological effects. Herein, on the ground of pre-clinical findings we unravel the underlying
mechanisms behind the plausible contribution of HMGB1 and its receptors (RAGE and TLR4) in the ALS pa-
thogenesis. Furthermore, we provide an account of the therapeutic outcomes associated with inhibition/blocking
of HMGB1 receptor signalling in preventing motor neuron’s death and delaying disease progression in ALS
experimental models. There is strong evidence that HMGB1, RAGE and TLR4 signaling axes might present po-
tential targets against ALS, opening a novel headway in ALS research that could plausibly bridge the current
treatment gap.

1. Introduction total population worldwide, and the median survival of ALS patients is
estimated around 24 months in Europe. Despite extensive research ef-
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegen- forts towards the elucidation of its pathogenesis, no disease-modifying
erative disease also known as Lou Gehrig’s disease, characterized by treatment is available to date [3].
rapid degeneration of motor neurons particularly in anterior horns of ALS presents a multifaceted pathophysiology reflecting a complex
the spinal cord and brainstem, as well as of large pyramidal neurons interplay between genetic and environmental factors. The vast majority
mainly in the primary motor cortex [1,2]. Epidemiological data have (90 %) of cases are “sporadic” occurring without a known cause or a
shown that the current prevalence of ALS is about 5 per 100,000 of the family history, whereas 5–10 % of them are familial-type ALS (FALS)

Abbreviations: HMGB1, high mobility group box 1; RAGE, receptor for advanced glycation end products; TLRs, toll-like receptors; ALS, amyotrophic lateral sclerosis;
FALS, familial ALS; SALS, sporadic ALS; LPS, lipopolysaccharide; fr-HMGB1, fully reduced HMGB1; ds-HMGB1, disulfide HMGB1; ox-HMGB1, sulphonyl HMGB1;
SOD1, Cu2+/Zn2+ superoxide dismutase 1; mSOD1, Mutant SOD1; mSOD1G93A, G93A mutant form of SOD1; SOD1WT, wild‐type SOD1; WT, wild-type; DAMP,
damage-associated molecular patterns; ERK, extracellular signal-regulated kinase; STAT3, signal transducer and activator of transcription 3; AGER, Advanced
glycosylation end‐product specific receptor; PRRs, pathogen recognition receptors; MHC, major histocompatibility complex; BBB, blood-brain barrier; CNS, central
nervous system; DCs, dendritic cells; GFAP, glial fibrillary acidic protein; IL, interleukin; IBA1, ionized calcium-binding adapter molecule 1; iNOS, inducible nitric
oxide synthase; NADPH, nicotinamide-adenine dinucleotide phosphate; NF-κB, nuclear factor κ light chain enhancer of activated B cells; TNF-α, tumor necrosis
factor-α; ChAT, anti-choline acetyltransferase; CD, cluster of differentiation; BDNF, brain-derived neurotrophic factor; GDNF, glial cell line-derived neurotrophic
factor; IR, immunoreactivity

Corresponding authors at: Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan
Lagoon Selatan, Bandar Sunway, Selangor, Malaysia.
⁎⁎
Corresponding author at: Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75M. Asias Street, 11527, Athens,
Greece.
E-mail addresses: paudel@monash.edu (Y.N. Paudel), cpiperi@med.uoa.gr (C. Piperi), farooq.shaikh@monash.edu (M.F. Shaikh).

https://doi.org/10.1016/j.phrs.2020.104792
Received 23 July 2019; Received in revised form 14 February 2020; Accepted 1 April 2020
Available online 08 April 2020
1043-6618/ © 2020 Elsevier Ltd. All rights reserved.
Y.N. Paudel, et al. Pharmacological Research 156 (2020) 104792

associated to genetic mutations which are typically autosomal dom- multi-ligand receptor belonging to the immunoglobulin (Ig) super-
inantly inherited [4]. The most common genetic causes of FALS involve family with a molecular weight of 45 kDa [26]. RAGE is composed of
mutations in the genes encoding for superoxide dismutase 1 (SOD1) (20 three Ig domains (V-type, C1-type and C2-type), a transmembrane do-
%), TAR DNA-binding protein 43 (TDP-43) (1–5 %), chromosome 9 main, and an intracellular cytoplasmic domain [27]. RAGE is mainly
open reading frame 72 (C9orf72) (40 %) and fused in sarcoma (FUS) expressed in endothelial cells, vascular smooth muscle cells, neurons
(1–5 %) [5]. These gene mutations disrupt the degradation of ag- and macrophages/monocytes [28]. RAGE gene is situated on chromo-
gregated proteins, impair central nervous system (CNS) glial protective some 6 of the major histocompatibility complex (MHC) class III region,
responses, and promote pro-inflammatory-mediated motor neuron in- which contains several other genes crucial for the adaptive and innate
jury [6]. Research advances in ALS genetics and molecular pathogen- immune responses [29].
esis, indicate that ALS is rather a syndrome and not a single disorder RAGE has been recognized as a multi-ligand receptor that can bind
[7]. Although the exact etiology of ALS is still largely unknown, accu- to HMGB1, S100, amyloid-β peptide, DNA, RNA, and other molecules
mulating evidence suggests that various cellular and systemic pro- that regulate several physiological processes [30]. RAGE signaling is
cesses, such as innate immune system, neuroinflammation, systemic upregulated under conditions of chronic and sustained inflammation
inflammation, oxidative stress, mitochondrial dysfunction, and ex- and has emerged as a key regulator of the innate immune response..
citotoxicity contribute to its pathogenesis [1,7,8]. Notably, the pathogenic role of RAGE in mediating inflammatory
High mobility group box 1 (HMGB1) acts as a pro-inflammatory neurodegeneration has been well-documented [31]. To date, studies
cytokine, as well as an initiator and amplifier of neuroinflammation have been mainly focused on the HMGB1/RAGE-induced nuclear factor
that exerts its biological activity mainly through the activation of its kappa-light-chain-enhancer of activated B cells (NF-κB) activation and
principal receptors, the receptor for advanced glycation end products the associated inflammatory responses [32]. Moreover, RAGE protein
(RAGE) and toll-like receptor 4 (TLR4) [9]. An increasing body of expression has been found upregulated in brain tissues of patients with
evidence suggests that HMGB1 is implicated in several CNS disorders inflammatory neurodegenerative conditions and disorders, including
such as Parkinson’s disease (PD) [10,11], Alzheimer's disease (AD) AD [33], PD [34] and ALS [35]. Accumulating evidence suggests that
[12], Epileptogenesis [13,14], Multiple sclerosis (MS) [15,16] and ALS RAGE activation, driven by increased availability of its ligands, could
[17] mainly by activation of RAGE/TLR4 signalling axes. also contribute to ALS pathogenesis [31].
RAGE and TLR4 are two main components of the innate immune The other HMGB1 receptor, TLR4 is the main member of the TLR
system and HMGB1 acts as an endogenous ligand for both of them [18]. family that responds to bacterial LPS, an essential component of the
Pre-clinical evidence has shown that HMGB1/RAGE and HMGB1/TLR4 outer membrane of gram-negative bacteria [36]. Ten functional TLRs
signalling axes are upregulated in ALS, resulting in motor neuron injury (TLR1-10) have been identified in humans, which can be divided into
and progression of inflammation [19], suggesting that HMGB1 may two groups; extracellular TLRs that recognize microbial membrane
play a pivotal pathogenic role in ALS progression. In this review, we components (TLR1, 2, 4, 5, 6, and 10), and intracellular TLRs that re-
provide an update of pre-clinical studies investigating the role of cognize microbial nucleic acids (TLR 3, 7, 8, and 9) [37]. TLR4, LPS-
HMGB1, RAGE, and TLR4 in mediating neuroinflammation and disease binding protein (LBP), cluster differentiation antigen 14 (CD14) and
progression in ALS. Furthermore, we discuss the therapeutic potential myeloid differentiation protein (MD-2) mediate the induction of in-
of HMGB1, RAGE, and TLR4 inhibition/blockade against animal models flammatory responses triggered by LPS. In general, LBP binds to LPS
of ALS pathogenesis along with future research directions. aggregates leading to the formation of an activated (TLR4/MD-2/LPS)
complex that plays a crucial role in initiating the inflammatory cascade
2. HMGB1 interaction with RAGE and TLR4 receptors [38]. TLR4-HMGB1 interaction triggers cytokine production. The
TLR4/MD-2 complex is required for HMGB1-mediated cytokine pro-
HMGB1 is the most abundant member of the HMGB family which duction in macrophages, with ds-HMGB1 being mandatory for this in-
consists of HMGB1-4 proteins [20]. HMGB1 is a ubiquitous, highly teraction [39,40]. The binding of ds-HMGB1 redox isoform to the TLR4
conserved nuclear protein with a molecular weight of 25-kDa, con- co-receptor MD-2 resembles LPS binding, but it occurs at a different
sisting of 215 amino acid residues. It is organized in two DNA-binding position [41]. Notably, TLR4, and its co-receptor CD14 have been
domains (termed A and B-boxes) and a negatively charged C-terminal considered as key players of the neuroimmune dysregulation associated
tail that contains a repeating chain of 30 continuous glutamic and as- with ALS [19]. Mutant SOD1 (mSOD1) has been reported to bind to
partic acids [21]. CD14 in ALS, which is a co-receptor of TLR4. Interestingly, microglial
HMGB1 is a prototypical alarmin and a pleiotropic danger-asso- activation mediated by mutant SOD1 can be attenuated using TLR4 and
ciated molecular pattern (DAMP) molecule, that is passively released by CD14 blocking antibodies [42].
damaged cells or secreted from activated immune cells into the extra- Among the 14 different receptors of HMGB1 identified to date
cellular space. HMGB1 can be secreted by activated monocytes, mac- [30,43], RAGE and TLR4 are the most extensively studied in HMGB1-
rophages, and microglial cells in response to several stimuli like lipo- mediated pathologies. Extracellular HMGB1 can interact with several
polysaccharide (LPS), tumour necrosis factor-α (TNF-α) or interleukin- pathogen recognition receptors (PRRs) to promote cell migration and
1β (IL-1β) [22–24]. In turn, secreted HMGB1 can amplify the release of cytokine production. The ability of HMGB1 to interact with multiple
several cytokines from inflammatory cells and stimulate the release of unrelated receptors in order to induce diverse functions is tightly
HMGB1 from the same cells that activate and maintain the in- regulated by post-translational redox modifications of the three cy-
flammatory cascade [22,23]. steine residues at positions 23, 45, and 106 in the box A and B domains
Moreover, HMGB1 has three isoforms with distinct functions [25]. [21]. Although all redox forms of HMGB1 can bind to RAGE, only ds-
The first one is fully reduced HMGB1 (fr-HMGB1), consisting of a thiol HMGB1 isoforms bind with high affinity [44]. In particular, a struc-
group in each of the cysteine residues that induces cell migration. The tural-functional study reported that a specific region consisting of the
second is the disulfide HMGB1 (ds-HMGB1) containing a disulfide amino acids 150-183 in the C terminal of HMGB1 is responsible for
bridge between C23 and C45 and a reduced C106 residue, being a TLR4 RAGE binding [45].
ligand. The third isoform is sulphonyl HMGB1 (ox-HMGB1), with
terminally oxidized cysteine residues and no pro-inflammatory activity, 3. Potential role of HMGB1 in ALS pathology
which can induce cell migration or cytokine production [21]. Among
the three isoforms, ds-HMGB1 is the only one contributing to in- Before moving to HMGB1, it is worth briefly discussing the role of
flammatory processes [21]. neuroinflammation in the ALS pathogenesis, which has gained in-
RAGE is a type 1 transmembrane glycoprotein, cell surface and creased attention. Neuroinflammation, mainly characterized by

2
Y.N. Paudel, et al. Pharmacological Research 156 (2020) 104792

reactive astrocytes and microglia, infiltration of peripheral immune and SOD1WT astrocytes demonstrated upregulation of RAGE and TLR4,
cells and upregulated inflammatory mediators, has been consistently not observed in SOD1G93A astrocytes, indicating that protective sig-
observed in motor regions of the CNS in both sporadic and FALS cases naling pathways amplified by extracellular ds-HMGB1 in astrocytes
[46]. However, microglia exert both neuroprotective as well as neuro- may be disrupted in ALS [17].
toxic effects depending on the stage of the disease progression as well as In addition to the SOD1G93A transgenic mouse model, the mod-
on cytokine secretion, indicating that deciphering the role of neuroin- ulation of HMGB1 levels has been also observed in the Wobbler mouse
flammation in ALS is complex but rather promising [47]. model of motor neuron degeneration [55]. It is worth noting that,
HMGB1 can initiate a positive feedback loop, which has been shown Wobbler mice recapitulate all the clinical phenotypes of human ALS
to aggravate the neuroinflammatory and neurodegenerative processes patients [56,57] as well as they exhibit ALS-typical symptoms such as
both in humans as well as in experimental ALS cases [19,48]. The pa- damage of the motor system [58]. Immunostaining for the HMGB1 and
thogenic role of HMGB1 in ALS has been unravelled from a study using its principal receptor (TLR4) revealed a significantly higher number of
a SOD1G93A transgenic mouse model (SOD1 mutant with a Gly-93-Ala immunopositive cells in diseased Wobbler mice compared to normal
substitution), a well-established animal model of FALS [48]. Increased controls [55]. This upregulation of HMGB1 and TLR4 supports the
HMGB1 immunoreactivity (IR) was observed mainly in the cell body notion that neurodegeneration and neuroinflammation co-occur in ALS.
and proximal neurites of labelled motor neurons with no differences It is worth noting that the correlation between HMGB1 and disease
however, been reported between controls and pre-symptomatic progression cannot be completely ascertained as the upregulation in the
SOD1G93A mice. Extracellular release of HMGB1 was associated with HMGB1 levels might be attributed both to neuronal death as well as to
disease progression, stimulated by cellular damage, thus reflecting the disease progression. It is highly possible that the degenerating motor
potential of HMGB1 to act as an extracellular inflammatory signal. neurons as well as the associated neuroinflammatory process, induces
Upon ALS progression, there was a reduction in HMGB1 IR by degen- HMGB1 release from the activated astrocytes and microglia in
erating neurons indicating the extracellular release of HMGB1 [48]. In hSOD1G93A mice. Hence, HMGB1 signaling may contribute to the pro-
accordance, another study showed an increased mRNA expression of inflammatory reactions exacerbating disease progression in ALS [49].
HMGB1 in the lumbar spinal cord of human mutant (h) SOD1G93A HMGB1 plausibly contributes to the progressive inflammatory and
transgenic mice during key disease stages. HMGB1 mRNA levels were neurodegenerative events in response to neurotoxic settings in the
elevated (1.7- fold) at the end-stage of disease, when compared with spinal cord of SOD1G93A mice, rather than presenting a primary event
wild type (WT) mice. At the protein level, HMGB1 expression was in- in the motor neuron death. However, in motor neurons undergoing
creased at the end-stage of disease by 2.5 fold [49]. Regarding the lo- degeneration, a downregulation of HMGB1 levels might be a con-
calization pattern of HMGB1 in these models, HMGB1 exhibited diffuse sequence of its passive release upon cellular damage, thus acting as an
nuclear staining and was mainly co-localised with CD11B-labelled mi- inflammatory signal [48].
croglia and glial fibrillary acidic protein (GFAP)-positive astrocytes in HMGB1 IR in the cervical spinal cord of control patients was mainly
WT mice, whereas no co-localisation with anti-choline acetyltransferase observed in the cytoplasm of motor neurons, whereas in ALS cases,
(ChAT)-positive motor neurons was observed. However, in hSOD1G93A residual motor neurons demonstrated variable nuclear/cytoplasmic IR.
mice, HMGB1 immunolabelling displayed increased intensity in GFAP- Resting glial cells showed nuclear expression in control patients,
positive astrocytes and CD11b-labelled microglia. Similar to WT mice, whereas cytoplasmic HMGB1 staining was substantially increased in
there was no HMGB1 immunolabelling observed in ChAT-positive the glial cells of ALS patients [19]. Interestingly, HMGB1 expression
motor neurons of hSOD1G93A mice [49], where ChAT, CD11b, and levels were confined in glial cells, including both astrocytes and acti-
GFAP act as cellular markers for motor neurons, microglia and astro- vated microglial cells. Quantification of HMGB1-positive glial cells
cytes, respectively. These findings support the pathogenic role of confirmed the increased cytoplasmic HMGB1 staining in ALS, as com-
HMGB1 in ALS, suggesting that HMGB1 may be a key player in the pro- pared to control spinal cord specimens [19]. These findings indicate an
inflammatory processes that exacerbate disease progression in ALS. The altered subcellular localization of HMGB1 in ALS, supporting the notion
observed nuclear to cytoplasmic translocation of HMGB1 in the reactive that it could be significantly involved in ALS pathogenesis, possibly due
astrocytes and microglia in ALS patients as well as animal models in- to its pro-inflammatory effects.
dicates the plausible pathogenic role for HMGB1 in ALS [19,48,49].
It is of no surprise that astrocytes are highly implicated in neuro-
degeneration and disease progression in ALS [50]. They can be toxic to 3.1. HMGB1 as a potential biomarker of ALS
motor neurons through the release of neurodamaging molecules
[51,52] or may indirectly contribute to motor neuron degeneration The notion of HMGB1 as a potential biomarker of ALS has been
through loss of their homeostatic and neurosupportive functions strengthened due to the increased extracellular HMGB1 release in
[53,54]. HMGB1-mediated signaling results in an astrocytic production plasma/serum and tissue during ALS pathology.
of neurotrophic factors in normal spinal cord astrocytes, namely brain- Findings from human sporadic ALS and FALS patients reported
derived neurotrophic factor (BDNF) and glial cell line-derived neuro- upregulated levels of HMGB1 autoantibodies in the serum of ALS pa-
trophic factor (GDNF), whereas dysregulation of astrocytic HMGB1 tients as compared to age-matched healthy controls. Intriguingly, the
signaling has been implicated in ALS [17]. Particularly, ALS motor cells concentration of anti-HMGB1 autoantibodies was 1.4-fold higher in
exhibited a nuclear to cytoplasmic translocation of HMGB1 with time, female ALS patients compared to male. However, no associations were
suggesting that HMGB1 could be also gradually released into extra- detected between anti-HMGB1 autoantibodies levels and onset types,
cellular milieu during disease progression. On the other hand, non- ALS subtypes, either familiar or sporadic [59].
transgenic (non-Tg) and SOD1WT motor neurons exhibited only a basal Similarly, another study that used post-mortem tissues obtained
nucleus-to-cytoplasm shift of HMGB1 [17]. The potential implication of from 12 sporadic ALS patients demonstrated upregulated HMGB1
HMGB1 signaling in motor neuron-astrocyte interaction in ALS was mRNA expression in ALS-long term patients (slow disease progression
further investigated using primary spinal cord astrocytes from non-Tg, with long-term survival >48 months) compared to control spinal cord
SOD1WT and SOD1G93A transgenic mice exposed to ds-HMGB1 at tissues. In accordance, ALS-short term patients (rapid disease progres-
asymptomatic stage (30 days), the onset of motor deficits (100 days) sion with short-term survival <18 months) displayed similar elevated
and at symptomatic stage (130 days). In this study, non-Tg and SOD1WT HMGB1 levels [19]. These findings suggest that HMGB1 auto Abs could
astrocytes responded to the increased BDNF and GDNF expression, serve as potential clinical biomarkers for ALS, in monitoring disease
which was not observed in SOD1G93A astrocytes. Interestingly, non-Tg progression.

3
Y.N. Paudel, et al. Pharmacological Research 156 (2020) 104792

4. The role of RAGE in ALS pathogenesis: Mechanistic insights might accelerate neurodegeneration and could also be a risk factor for
from experimental and clinical studies ALS [68]. Elevated expression of S100β has been also detected in the
serum [69,70], as well as in rat motor neurons exposed to cerebrospinal
In the nervous system, the precise role of RAGE is not yet fully fluid (CSF) of ALS patients [70]. This finding reflects the possible
elucidated. However, accumulating evidence demonstrates that RAGE contribution of S100β (RAGE ligand) in ALS and suggest that S100β
exerts its biological activities mainly via its implication in partially might be a potential biomarker [71].
cross-linked signaling pathways including extracellular signal-regulated However, it is not well understood whether RAGE interactions in
kinase (ERK), Janus kinase 2 (JAK2)-signal transducer and activator of the spinal cord mediate damage by promoting inflammation, oxidative
transcription 3 (STAT3) and NF-κB, [60]. RAGE is implicated in an stress or other molecular mechanisms. It is worth investigating whether
array of cellular neuronal processes, such as neuroinflammation, mu- RAGE and its ligands are the cause, or the modifiers of neurodegen-
tant protein aggregation, production of reactive oxygen species (ROS) erative diseases. Nevertheless, the upregulation of RAGE and its ligands
and AGE accumulation [61], contributing to neurodegeneration as well might present both a potential mechanism or a biomarker [35].
as to the development of neurodegenerative diseases, including ALS.
There is evidence that RAGE-mediated signaling leads to the increased 5. Implication of TLR4 signaling in ALS pathogenesis
synthesis and release of pro-inflammatory molecules and elevation of
nicotinamide-adenine dinucleotide phosphate (NADPH)-dependent TLRs, similar to RAGE, are considered as PRRs and their signaling
ROS production, resulting in the aggravation of neuroinflammation and activates transcriptional factors, cytokine synthesis, and secretion, as
oxidative stress, respectively. These mechanisms exacerbate the pa- well as promotes inflammatory reactions and brings macrophages and
thological processes that lead progressively to neuronal death [62]. neutrophils at the site of inflammation through gradient formation
Various cell types such as microglia, peripheral monocytes/macro- [62]. TLR4, a member of the TLR family, is one of the most crucial
phages, astrocytes, T and B lymphocytes are supposed to contribute to receptors of innate immunity and is widely expressed by immune and
ALS pathology. Interestingly, RAGE is expressed in many of these cell nonimmune cells, including CNS-resident immune cells [72,73]. TLR4
types and its effects have been studied in both pre-clinical and experi- signaling axis can activate two main downstream pathways, one
mental models of ALS [63]. MyD88-dependent and MyD88-independent, leading to the activation
Experimental in vivo evidence has shed light on the contribution of of NF-κB pathway which in turns activates downstream cytokines, such
RAGE in ALS pathogenesis. In particular, RAGE and mRNA expression as NO, TNF-α, and IL-1β [74]. Activated TLR4 is suggested to promote
of advanced glycation end product-specific receptor (AGER) levels were the secretion of many cytokines ultimately aggravating the neuroin-
found increased (by 2.24 folds) in lumbar spinal cord specimens of flammatory mechanisms that are involved in ALS [75].
SOD1 transgenic mice in comparison to controls at the end-stage of the Glial TLR4 signalling has been implicated in disease progression and
disease. This evidence suggests that RAGE contributes at least in part, to contributes to end-stage ALS pathology in hSOD1G93A mice.
the chronic as well as the constant neuronal loss observed in this dis- Expression levels of TLR4, along with the respective mRNAs, were
order [64]. shown to be upregulated in microglial and astrocytic cells of mutant
The putative role of RAGE in ALS has been also investigated in vitro. SOD1 mice as compared to WT controls. Interestingly, a prolonged
The Nerve growth factor (NGF) protein has been found to be post- disease course and a better motor performance were demonstrated in
translationally modified by oxidation and contribute to RAGE signaling- transgenic mice lacking TLR4, implicating TLR4 as a notable player in
mediated motor neuron death, when normal motor neurons are co- motor neuron degeneration [49].
cultured with SOD1 G93A astrocytes. in vitro glycation of NGF has been Extracellular mSOD1 has been shown to cause motor neuron injury
shown to favour oligomerization and lead to the generation of RAGE and trigger microgliosis in spinal cord cultures [42]. However, it has
ligand and modified NGF, further inducing motor neuron death and been unclear whether extracellular mSOD1 can directly interact with
astrocyte-mediated motor neuron toxicity [65]. When exosomes from motor neurons themselves or whether mSOD1‐activated microglia
mutant SOD1-transfected NSC-34 cells were incubated with N9 micro- mainly contribute to the extracellular mSOD1-mediated motor neuron
glia-like cells, there was an increased upregulation of RAGE, TLR4, IL- neurodegeneration. It has been reported that mSOD1‐associated neu-
10, and TREM as compared to controls [66]. Moreover, S100β (RAGE rotoxic mechanisms are mediated by microglia and the activation of the
ligand) expression was significantly elevated in C6 rat astrocytoma cells TLR/CD14 pathway involves TLR4 and TLR2 [42], indicating a striking
overexpressing mutant SOD1 G93A protein [67]. link between mSOD1-mediated microglial activation and neurodegen-
A clinical study provides a further molecular glimpse into the con- eration. In addition, granulocyte macrophage-colony stimulating factor
tribution of the RAGE/AGE axis in ALS. Quantitative analysis showed (GM-CSF), a pleiotropic cytokine predominantly released by astrocytes
increased RAGE expression, mRNA expression of AGER (the gene en- that up-regulates TLR4 and CD14 expression in microglia, exacerbate
coding RAGE) and protein expression of RAGE as determined by TLR signaling in ALS [76].
Western blot analysis in the spinal cords of ALS patients when com- Upregulated levels of TLR4 and NF-κB has been observed in a
pared to controls, suggesting the possible implication of RAGE pathway transgenic model of ALS (SOD1G93A). Particularly, increased TLR4
in ALS. These findings imply that selective loss of motor neurons may expression and nuclear protein levels of NF-κB was observed in the
be at least partially attributed to the regulation of the RAGE/AGE brainstem of SOD1G93A rats compared to WT animals [77]. The acti-
pathway in ALS [35]. An upregulation of RAGE expression in reactive vation of the TLR4/NF-κB cascade is a priming signal that induces the
glial cells in both grey and white matter of the spinal cord was de- transcription of inflammasome components. It is known that TLR4 in-
monstrated in human sporadic ALS specimens. RAGE was expressed in itiates an activation of NF-κB, leading to the activation of the tran-
reactive astrocytes (GFAP positive cells) and cells of the microglial/ scription of pro-inflammatory cytokines [78]. This finding highlights
macrophage lineage (HLA-DR positive cells) while a higher RAGE glial the crucial role of neuroinflammation in ALS.
IR score was reported in both ALS-short-term and ALS-long-term pa- Clinical findings have also shed more light on the plausible con-
tients as compared to controls [19]. These findings provide strong tribution of TLR4 in ALS pathogenesis. More specifically, elevated TLR4
evidence regarding the possible role of RAGE signalling in the ALS mRNA expression levels were observed in the spinal cord of both
pathogenesis. sporadic ALS-short-term and ALS-long-term patients, as compared to
Soluble RAGE (sRAGE), another potential modulator of inflamma- controls. Regarding the cellular distribution of TLR4 in ALS, its staining
tion, has been found reduced in the serum of ALS patients. However, no intensity was elevated in cells with typical glial morphology in both
positive correlation between the serum sRAGE levels and clinical white and grey matter, whereas moderate TLR4 IR was detected in
parameters of the disease was obtained. This study suggests that sRAGE control motor neurons. TLR4 expression was further observed in glial

4
Y.N. Paudel, et al.

Table 1
Representative studies reporting HMGB1, RAGE and TLR4 targeted therapies in ALS.
S.N. Interventions Model Treatment schedule Observations References

G93A
1 Anti-HMGB1 antibody (2G7) SOD1 transgenic mouse • For early treatment-administered weekly. • Pre-onset treatment with anti-HMGB1 antibody improved hind-limb grip [1]
(100 μg/mouse injection, I.P.) (C57BL/6 J) model of ALS • For therapeutic treatment- administered 70 days postnatal strength but did not extend survival.
and continued weekly throughout the end stage of the treatment with anti-HMGB1 antibody does not extend survival or
G93A
diseases.
• Post-onset
improved motor performance in SOD1 mice.
• Another cohort of animals received anti-HMGB1 antibody • Treatment with anti-HMGB1 antibody reduced TNF-α and complement C5aR1
from 35 days till 133 days postnatal. gene expression in the spinal cord but does not modulate the activation of glial
cell.
2 Soluble RAGE (natural competitor SOD1 transgenic mice model of • Soluble RAGE (175 μg/day I.P.) starting at 8 weeks of age • Soluble RAGE-treated mice demonstrated prolonged life span (138.5 ± 3.1 vs. [2]
of RAGE)(175 μg/day, I.P.) ALS until end of an experiment. 130.6 ± 3.1 days), delays progression of ALS and improved functional
performance as compared to murine serum albumin (MSA) treated group.
a Soluble RAGE-treated SOD1 mice, higher neuronal counts and lower
• Inastrocytes has been observed.

5
3 TLR4 inhibitor (TAK-242) Transgenic hSOD1G93A mice TAK-242 (3 mg/kg, I.P.), three times per week was treatment (n = 13) decreases the clinical disease progression in [3]
G93A
model of ALS administered to hSOD1G93A mice starting at the age of 7 week.
• TAK-242
hSOD1 mice compared with vehicle treated (n = 16) group as evident by
behavioral motor performance analysis (ladder score, hind limb score and
percent survival)
• TAK-242 treatment attenuated motor neuron loss in the spinal cord of
hSOD1G93A mice as well as attenuated spinal cord astrogliosis and microglia
activation in hSOD1G93A mice.
4 TLR4 antagonist In vitro model of ALS – • LPS (1 μg/mL) induced motor neuron death was countered by TLR4 [4]
(IAXO102 and FP7) antagonist. IAXO102 (10 μM) and FP7 pre-treatment 2 h before LPS exposure
significantly reduce motor neuron death.
5 TLR4 antagonism Transgenic hSOD1G93A mice TLR4−/− female C57BL/6 mice were bred with male • TLR4 lacking mice (hSOD1G93A × TLR4−/−) improves hind-limb grip [5]
model of ALS hSOD1G93A to yield hSOD1G93A mice lacking TLR4 (hSOD1G93A strength at 56, 63, 84 and 161 days of age (n = 8–9, *P < 0.05) and extends
× TLR4−/−). survival when compared to hSOD1G93A mice (184 vs 169, n = 11, *P < 0.05)

HMGB1, High mobility group box 1; ALS, amyotrophic lateral sclerosis; RAGE, Receptor for advanced glycation end products; TLR4, Toll-like receptor 4; SOD1, Cu2+/Zn2+ superoxide dismutase 1; LPS,
Lipopolysaccharide; TNF-α, Tumor necrosis factor-α, I.P., Intraperitoneal; C5Ar1, Complement component 5a receptor 1.
Pharmacological Research 156 (2020) 104792
Y.N. Paudel, et al. Pharmacological Research 156 (2020) 104792

cells of ALS specimens, but not in resting glial cells, depicting the ex- 6.2. RAGE inhibition in ALS experimental models
pression pattern of the protein [19]. These findings strikingly suggest
that the activation of TLR4 axis might demonstrate crucial role in Despite the increasing evidence that implicates the contribution of
progression of inflammation, resulting in progressive degeneration of RAGE in ALS pathogenesis, experimental studies evaluating the ther-
motor neurons in ALS [19]. apeutic potential of RAGE inhibition against ALS are still limited.
These experimental and clinical findings suggest that TLR4 plays a However, a previous study has reported the significance of RAGE
crucial role in the progressive motor neuron degeneration in ALS, po- blockade by sRAGE (175 μg/day, I.P.) in SOD1G93A mutant mice [64].
tentially reflecting the significant implication of the innate immune sRAGE presents a soluble form of RAGE that serves as a natural in-
system in sensing neuronal injury and driving disease progression [79]. hibitor, suppressing RAGE function, sequestering RAGE ligands and
blocking their interaction with cell surface RAGE, thus preventing their
6. Potential impact of HMGB1, RAGE, and TLR4 inhibition in ALS subsequent harmful effects on effector cells [81]. sRAGE has been re-
therapy ported to delay disease progression in ALS and was also shown to
prolong the life span of SOD1 transgenic mice, compared to controls
Since there is no disease-modifying treatment available for ALS to date, [64]. sRAGE administered to male SOD1G93A mutant mice (starting at
current research has been focused on the exploration of novel effective 8 weeks until sacrifice) has been shown to extend the life span and
therapeutic alternatives. Based on the evidence of HMGB1, RAGE and delay the progression of ALS symptomatology [64]. Mice treated with
TLR4 contribution to ALS pathogenesis, their inhibition/modulation re- sRAGE exhibited a higher probability of surviving for a long time after
presents a possible therapeutic alternative approach (Table 1). the onset of the disease and showed a relatively longer lifespan
(138.5 ± 3.1 vs. 130.6 ± 3.1 days), compared to the murine serum
6.1. HMGB1 blockade in ALS experimental models albumin (MSA)-treated group. Moreover, sRAGE-treated mice demon-
strated a higher number of neurons at the end stage of the disease,
Recent advances in HMGB1 biology and its release pattern in the compared to the control group. On the contrary, GFAP-im-
pathophysiology of several diseases have revealed HMGB1 as a pro- munoreactivity was lower in the spinal cord of sRAGE-treated mice as
mising treatment target. A growing body of evidence indicates the compared to the MSA-treated ones, reflecting higher neuronal survival
putative role of HMGB1 and its receptor signalling (RAGE/TLR4) in ALS and lower astrocytosis in sRAGE-treated SOD1 mice [64].
pathogenesis. However; very few experimental studies have in- Despite the promising effects of RAGE inhibition, the underlying
vestigated the therapeutic potential of HMGB1 blockade against ALS. mechanism of sRAGE-mediated therapeutic effects in ALS remain un-
In this context, Lee et al. have demonstrated that the therapeutic ravelled and the distinct time courses for initiation of sRAGE adminis-
blockade of HMGB1 before and after disease onset by intraperitoneal (I.P.) tration were not tested [63]. Nevertheless, the results of this study
administration of anti-HMGB1 antibody (2G7) (100 μg/mouse injection, suggest that sRAGE or other forms of RAGE blockade/inhibition might
I.P.) reduces early motor deficits in SOD1G93A transgenic mice [18]. represent an alternative therapeutic strategy against ALS.
Pharmacological blockade of HMGB1 was found to ameliorate hind-limb
grip strength but did not extend survival or improved motor performance 6.3. TLR4 inhibition in ALS experimental models
as compared to control antibody-treated SOD1G93A mice. Notably, the
transcripts of Itgam, CD68, Aif1 which are markers of microglia and in- As discussed above, TLRs (TLR4, TLR2 and their co-receptor CD14)
filtrating monocyte/macrophages as well as the mRNA expression levels of and especially TLR4 may be highly implicated in ALS pathogenesis. For
astrocytic marker, GFAP remained unchanged. On the other hand, tran- instance, TLR4 signaling in pro-inflammatory microglial cells has been
scripts of Ly6c which is a predominant marker of early infiltrating implicated in the degeneration of motor neurons, leading to ALS pa-
monocytes/macrophages in lumbar spinal cord were found reduced in thology [82]. Therefore, targeting TLRs has received increasing atten-
anti-HMGB1 antibody-treated SOD1G93A mice, as compared to control tion as a potential novel treatment strategy against ALS.
antibody-treated SOD1G93A mice. These results suggest that anti-HMGB1 TAK-242 (3 mg/kg, I.P.), a specific TLR4 inhibitor has been reported
antibody treatment does not alter microglia and astrocytes. to delay disease progression, attenuate motor neuron loss and decrease
In addition, treatment with anti-HMGB1 antibody reduced mono- astrogliosis and microglial activation in the spinal cord of hSOD1G93A
cyte markers in the tibialis anterior (TA) and gastrocnemius (GN) as mice. These effects were accompanied by attenuation of disease pro-
assessed by the unaltered mRNA expression of macrophage markers gression as well as motor improvement, when assessed by motor be-
(Itgam, CD68 and Aif1), and decreased mRNA expression of monocyte havioral and ladder testing in TAK-242-treated hSOD1G93A mice. TAK-
marker (Ly6c) in anti-HMGB1 antibody treatment group compared to 242-treated hSOD1G93A mice demonstrated the delayed deterioration
control mice [18]. TNF-α mRNA expression and C5a receptor 1 (C5ar1) of hind-limb reflex, as compared to vehicle-treated hSOD1G93A mice.
were significantly downregulated (0.27-fold and 0.22-fold respectively) TAK-242 also exerted immunomodulatory effects as shown by the re-
in the spinal cord of the anti-HMGB1 Ab treatment group, compared to duced serum IL-1β levels in TAK-242-treated hSOD1G93A mice [83].
control antibody-treated SOD1G93A mice, whereas mRNA expression TAK-242 treatment attenuated motor neuron loss and inhibited spinal
of IL-1β, AGER and TLR4 remained unaffected [18]. cord astrogliosis and microglia activation in the spinal cord of
However, therapeutic ablation of HMGB1 signalling protects against hSOD1G93A mice, as evidenced by a significant attenuation of GFAP
early motor deficits but does not extend survival time and demonstrates and ionized calcium-binding adapter molecule 1 (IBA1), respectively.
overall limited efficacy in the SOD1G93A mouse model of ALS. A possible Although TAK-242 treatment downregulated mRNA expression of TNF-
reason behind the limited efficacy of HMBG1 neutralization in this study α, IL-1β mRNA levels remained unaffected [83].
could be attributed to the presence of other endogenous ligands that On a limiting aspect, TAK-242 treatment was found to exert only
trigger TLR2, TLR4 and RAGE, and ultimately contribute to disease pa- moderate and temporary clinical delay in the disease progression
thology. Moreover, earlier findings have also reported the weak brain and without extending survival in treated animals. Despite of the promising
spinal cord penetrating ability of antibodies [80] that could potentially findings, the study did not evaluate the peripheral effects of TAK-242
alter the inhibitory effect of the compound on CNS-derived HMGB1 [18]. treatment on the innervation or neuromuscular junction (NMJ) as
Nevertheless, this study provides evidence suggesting that pharmacolo- peripheral denervation generally occurs prior to motor neuron death
gical blockade/inhibition of HMGB1 might represent a promising ther- and symptoms onset in ALS [84]. Hence, this process may have started
apeutic strategy against ALS. However, the limited number of relative before or around the initiation of TAK-242 therapy, thus possibly hin-
studies reflects the urgent need of exploring more HMGB1 targeting agents dering the beneficial impact of the treatment [83].Nevertheless, these
and evaluating their therapeutic potential against ALS. results suggest that TLR4 manipulation could offer disease-modifying

6
Y.N. Paudel, et al. Pharmacological Research 156 (2020) 104792

HMGB1, High mobility group box 1; ALS, amyotrophic lateral sclerosis; RAGE, Receptor for advanced glycation end products; TLR4, Toll-like receptor 4; SOD1, Cu2+/Zn2+ superoxide dismutase 1; LPS,
effects against ALS, inferring that targeting of the innate immune

References
system via TLR4 regulation could serve as an emerging therapeutic
strategy against ALS.

[1]

[2]

[3]

[4]

[2]

[1]

[3]
Interestingly, TLR4 deletion/absence provides an extended survival in
the hSOD1G93A mouse models of ALS, further confirming the fact that

lumbar spinal cord tissue from SOD1 transgenic mice reported elevated mRNA expression AGER (2.2-fold higher) compared to normal control.
ALS spinal cord tissue exhibited significantly higher protein levels of HMGB1 compared to control samples (n = 3, *P < 0.05), as determined by western

elevation of HMGB1 mRNA expression was observed in ALS patients with patients with slow disease progression and long-term survival >48

TLR4 mRNA expression was noted in both ALS patients with rapid disease progression and short-term survival <18 months (ALS-ST) and in ALS-
analysis reported elevated expression of RAGE in SOD1 transgenic mice compared to normal control during the end stage of the
upregulation (1.7-fold) in HMGB1 mRNA levels were observed at the end-stage of disease as compared with WT mice (n = 9, **P < 0.01).
TLR4 signalling might contribute to end-stage ALS pathology in
hSOD1G93A mice. hSOD1G93A × TLR4−/− mice demonstrated significant

mice was observed at onset (1.4-folds), mid-symptomatic (1.6-fold) and end-stage (5.6-fold),
extended survival as well as improvement in hind-limb grip strength, in

mRNA expression of AGER (gene encoding RAGE) was reported in the thoracic spinal cords of ALS subjects compared to controls.
comparison to hSOD1G93A mice (184 days vs 169 days, n = 11, *P <

LT patients (patients with slow disease progression and long-term survival > 48 months) as compared to control spinal cord (P < 0.05).
0.05). Nevertheless, survival extension and motor improvement upon TLR4
deletion is only moderate, suggesting that TLR4 is probably only one of

protein expression was upregulated (2.9-fold) at the end-stage of disease as compared with WT mice (n = 4, *P < 0.05).
several contributing factors in hSOD1G93A ALS pathogenesis [49].
In spinal cord primary cultures from SOD1 G93A mice (an in vitro ALS

protein expression was increased (2.5-fold) at the end-stage of disease compared to WT mice (n = 4, *P < 0.05).
model) (as well as in LPS-driven neurotoxicity model, small molecules
IAXO102 (10 μM), FP7 (1 and 10 μM) and extra-virgin olive oil (EVOO) (1
and 10 μg/mL) (TLR4 antagonist) have been assessed for their therapeutic
potential [82]. LPS (1 μg/mL)-induced motor neuron death was counter-
acted by TLR4 antagonists, as evidenced by reduced motor neuron death
upon EVOO treatment. Further pre-treatment of the co-cultures with

immunostaining was upregulated in ALS thoracic spinal cord as compared to control tissue.

increased expression of RAGE in human ALS spinal cord compared to control samples.
IAXO102. FP7 and EVOO significantly prevented the LPS-mediated IL-1β
release, whereas no significant effects were observed for all three TLR4
antagonists on TNF-α release. Pre-treatment with EVOO almost com-
pletely blocked the nitric oxide (NO) release by LPS-stimulated cultures,
whereas IAXO102 and FP7 were not effective. Notably, IAXO102, FP7,
and EVOO reduced the SOD1mut glia-driven motor neuron death [82].

respectively as compared to WT mice (n = 9, *P < 0.05 and ***P < 0.001).


These findings provided compelling evidence that natural TLR4 antago-
nists not only prevent the motor neuron death induced by LPS but also
effectively protect motor neurons from the toxicity of microglia carrying
the SOD1G93A mutation and need to be further confirmed in vivo.

months (LT) as compared to the control spinal cord (P < 0.05).


Similarly, neuroprotective effects of cyanobacteria-derived TLR4 an-
tagonist (VB3323) have been assessed in cultures of the spinal cord and in
an experimental model of motor neuron degeneration [85]. VB3323

G93A
(20 μg/mL) prevented the activation of microglia induced by LPS (1 μg/
mL) as assessed by Western blot analysis. VB3323 also significantly re-

increase in TLR4 mRNA in hSOD1


duced the LPS-dependent cytokine release in co-cultures of motor neurons
and glia, as demonstrated by the downregulation of TNF-α, IL-1β, and IL-
6, reflecting its anti-inflammatory activity. Furthermore, in a mouse model
of motor neuron degeneration (Wobbler mice), VB3323 (5 mg/kg/day,
I.P.) treatment improved their performance in behavioral scores (paw
abnormality degree and the forelegs grip-strength measurements) as
• Immunohistochemical

compared to the Riluzole-treated mouse group. Glial cell activation and


Representative studies reporting upregulation of HMGB1, RAGE and TLR4 in ALS.

TNF-α expression were also reduced in the cervical spinal cord of Wobbler
mice upon VB3323 treatment [85]. Moreover, TLR4 blocking inhibited
• Significant

• Significant

• Significant
• Significant
• Similarly,
• Increased

microglial activation mediated by mutant SOD1G93A. Furthermore,


Observations

diseases.

• Elevated
• blotting.
• HMGB1
• HMGB1

TNF‐α production in mSOD1G93A‐treated microglia was downregulated


• TLR4

Lipopolysaccharide; TNF-α, Tumor necrosis factor-α, WT, Wild-type.


upon administration of TLR4 and TLR2 blocking antibodies [42]. To sum
up, these experimental findings support the role of TLR4 in ALS patho-
genesis and suggest that TLR4 antagonists/inhibitors may be potential
mice model of

Transgenic hSOD1G93A mice model of

therapeutic candidates against motor neuron degeneration in ALS.


SOD1 transgenic mice model of ALS

7. Discussion and future perspectives

ALS is a sporadic multifactorial motor neuron disorder, caused by a


G93A

Human ALS subjects

Human ALS subjects

Human ALS subjects

Human ALS subjects

largely unknown interplay between environmental, genetic and epige-


Transgenic hSOD1

netic modifications [86]. However, the underlying mechanism of motor


neuron death in ALS is still elusive. Among several contributing factors,
Study type

the role of the innate immune system and neuroinflammation are the
most extensively studied [1,6,8].
ALS

ALS

Till date, Riluzole (anti-glutamatergic drug) is the only available


treatment option against ALS that extends survival for approximately 3
Increased levels

months [87]. However, pre-clinical studies investigating the role of


Riluzole in ALS present contradictory results with both beneficial ef-
HMGB1

HMGB1

HMGB1

RAGE

RAGE

TLR4

TLR4

fects in SOD1 transgenic mice models [88], or no effects in disease


progression [89]. These contradictory findings and the lack of effective
Table 2

treatment options against ALS portray the complexity of the disease and
S.N.

reflects the urgent need of developing alternative therapeutic strategies


2

7
Y.N. Paudel, et al. Pharmacological Research 156 (2020) 104792

that not only ameliorate motor symptoms but also improve survival. further reflects the need for more experimental studies evaluating the
Emerging findings demonstrate an increased expression of HMGB1, therapeutic potential of HMGB1 neutralizing agents, such as anti-
RAGE, and TLR4 in reactive glial cells in experimental and human ALS, HMGB1 antibodies, natural HMGB1 inhibitors (glycyrrhizin) [97], re-
implicating the contribution of HMGB1 and its receptor signalling combinant BoxA, Ethyl pyruvate and Salicylic acid against ALS. Of re-
(RAGE and TLR4) in the pathogenesis of the disease (Table 2).The levance, a potential future HMGB1-based therapeutic strategy could be
distribution and cellular localization of HMGB1and its receptors (RAGE the investigation of ds-HMGB1, a specific isoform of HMGB1 that is
and TLR4) in the spinal cord of a transgenic mouse model of ALS considered to primarily contribute to the inflammatory process [98].
strengthen the possibilities of an activation of HMGB1 signaling path- Similarly, different available RAGE inhibitors such as sRAGE and ex-
ways in ALS. HMGB1 signaling cascades leads to aggravation of neu- tracellular RAGE inhibitors such as TTP488 and derivatives, FPS-ZM1
roinflammation leading to enhanced synthesis and release of pro-in- [27] should be also evaluated therapeutically against ALS.
flammatory molecules worsening the existing pathological processes Despite the promising outcomes upon therapeutic blockade of
and resulting in the motor neuron injury in relation to ALS (Fig. 1). HMGB1 by anti-HMGB1 antibody in ALS, there is also evidence that
More importantly, upregulation of HMGB1, and its receptors does not anti-HMGB1 antibody treatment provides no beneficial effect on the
directly cause ALS, but they serve as a risk factors for ALS. The precise motor decline, the survival, the alteration of spinal cord glial numbers
understanding on the plausible link between HMGB1/RAGE/TLR4 or their activation profiles in SOD1G93A mice [18]. These reflect the
signalling in ALS, aggravation of inflammatory cascade and progressive further need of exploring the role of HMGB1 on neuroinflammation and
motor neuron degeneration might represent a novel therapeutic disease progression in ALS. An abnormal immune system response so-
strategy against ALS mainly by inhibiting neuroinflammation. This is lely cannot be the causative factor leading to motor neuron death, and
further supported by experimental studies evaluating the therapeutic additional factors must be required for disease initiation [1].

Fig. 1. HMGB1 signaling axis in ALS.


HMGB1 has been implicated in the disease progression in ALS
mainly via the interaction with its receptor RAGE and TLR4.
Increased levels of HMGB1, RAGE, AGER (RAGE gene), S100B
(RAGE ligands) and TLR4 have been detected in ALS, sug-
gesting that HMGB1 signaling plausibly contributes to ALS
pathology mainly via aggravating neuroinflammation leading
to motor neuron death. Inhibition/blockade of HMGB1 sig-
naling axis has been shown to delay disease progression, ex-
tend survival, attenuate motor neuron loss and reduce
monocyte markers along with early motor deficits. These
promising findings indicate that HMGB1 signaling axis de-
serves further exploration against ALS that could aid in de-
veloping novel therapeutic strategies.
ALS, Amyotrophic lateral sclerosis; HMGB1, High mobility group
box 1; RAGE, Receptor for advanced glycation end products;
TLRs, Toll-like receptors; AGER, Advanced glycosylation end‐-
product specific receptor; TNF, Tumor necrosis factor; C5ar1, C5a
receptor 1

role of HMGB1, RAGE, and TLR4 inhibition/deletion in transgenic ALS Beside the pro-inflammatory roles of HMGB1, not much is known
mice. HMGB1, RAGE and TLR4 inhibition/blockade significantly pre- about the initiation mechanism of HMGB1 translocation and release
vented motor neuron death, delayed disease progression, reduced as- during disease condition. However, mobilization and release of extra-
trogliosis and inhibited microglial activation, suggesting that targeting cellular HMGB1 occurs not only under pathological conditions but also
of HMGB1 and its receptors (RAGE and TLR4) might overcome the under several physiological situations. This reflects the need of better
current therapeutic gap, with limited however, supporting findings.. understanding of the HMGB1 release mechanisms that would eventually
The SOD1G93A transgenic mouse strain offers early onset monitoring shed lights on the various physiological roles of HMGB1 [99]. Design and
and rapid progression of the disease, presenting with gradual loss of motor validation of antagonists with greater HMGB1 specificity are crucial along
neurons and low variability of the phenotype on defined genetic back- with the development of antagonists targeting regions of HMGB1 inter-
grounds. In this regard, SOD1G93A transgenic strain represents the most action with its receptors, representing a future area of research. Hence,
used animal model for deciphering the early events in the neurodegen- HMGB1-based therapies with negligible off-target effects should be used to
erative process and understanding the pathogenesis of the disease, as well strengthen its value as a specific and effective therapeutic target [100].
as for testing therapeutics designed to halt the progression of ALS [90]. In conclusion, the current review sheds light on HMGB1 receptors
However, despite the knowledge that SOD gene mutation is associated signalling in the ALS pathogenesis and reveals the diagnostic and
with FALS, the exact way that mutant SOD1 specifically alters motor prognostic biomarker potential of HMGB1, RAGE and TLR4 in ALS,
neuron physiology is not completely understood [91]. ALS is an auto- indicating a new frontier of research.
somal-dominant disorder and all the findings discussed herein are solely
based on the SOD1G93A transgenic mouse model of ALS. Hence, it would
be interesting to investigate the therapeutic outcome of HMGB1, RAGE Author’s contribution
and TLR4 inhibition/blockade on other animal models of ALS [92].
In spite of the well-reported beneficial effects of HMGB1 neu- YNP conceived, carried out the literature review and drafted the
tralization strategy in a wide range of HMGB1-mediated pathologies manuscript. EA and CP provided critical inputs, revised and edited the
such as Epilepsy [93], MS [94], Traumatic brain injury (TBI) [95] and final version of the manuscript. MFS and IO revised the draft. All au-
Cognitive decline [96], there is only one study till date that has ex- thors read and approved the final manuscript.
plored the therapeutic role of anti-HMGB1 antibodies in ALS [18]. This

8
Y.N. Paudel, et al. Pharmacological Research 156 (2020) 104792

Funding sources the SOD1 G93A mouse model of amyotrophic lateral sclerosis, J.
Neuroinflammation 16 (1) (2019) 45.
[19] M. Casula, A. Iyer, W. Spliet, J. Anink, K. Steentjes, M. Sta, D. Troost, E. Aronica,
This research did not receive any specific grant from funding Toll-like receptor signaling in amyotrophic lateral sclerosis spinal cord tissue,
agencies in the public, commercial, or not-for-profit sectors. Neuroscience 179 (2011) 233–243.
[20] M. Štros, HMGB proteins: interactions with DNA and chromatin, Biochimica et
Biophysica Acta (BBA)-Gene Regulatory Mechanisms 1799 (1-2) (2010) 101–113.
Ethics approval and consent to participate [21] H. Aucott, J. Lundberg, H. Salo, L. Klevenvall, P. Damberg, L. Ottosson,
U. Andersson, S. Holmin, H.E. Harris, Neuroinflammation in response to in-
Not applicable. tracerebral injections of different HMGB1 redox isoforms, J. Innate Immun. 10 (3)
(2018) 215–227.
[22] U. Andersson, H. Wang, K. Palmblad, A.-C. Aveberger, O. Bloom, H. Erlandsson-
Declaration of Competing Interest Harris, A. Janson, R. Kokkola, M. Zhang, H. Yang, High mobility group 1 protein
(HMG-1) stimulates proinflammatory cytokine synthesis in human monocytes, J.
Exp. Med. 192 (4) (2000) 565–570.
The authors declare that there is no conflict of interest regarding [23] H. Wang, O. Bloom, M. Zhang, J.M. Vishnubhakat, M. Ombrellino, J. Che,
this work. A. Frazier, H. Yang, S. Ivanova, L. Borovikova, HMG-1 as a late mediator of en-
dotoxin lethality in mice, Science 285 (5425) (1999) 248–251.
[24] H. Erlandsson Harris, U. Andersson, Mini‐review: the nuclear protein HMGB1 as a
Acknowledgement proinflammatory mediator, Eur. J. Immunol. 34 (6) (2004) 1503–1512.
[25] Y.N. Paudel, E. Angelopoulou, C. Piperi, V.R. Balasubramaniam, I. Othman,
YNP would like to acknowledge Monash University Malaysia for M.F. Shaikh, Enlightening the role of high mobility group box 1 (HMGB1) in in-
flammation: updates on receptor signalling, Eur. J. Pharmacol. (2019) 172487.
supporting with Graduate Research Merit Scholarship. [26] A. Pinkas, K.H. Lee, P. Chen, M. Aschner, A.C. elegans, Model for the study of
RAGE-Related neurodegeneration, Neurotox. Res. 35 (1) (2019) 19–28.
Appendix A. Supplementary data [27] B.I. Hudson, M.E. Lippman, Targeting RAGE signaling in inflammatory disease,
Annu. Rev. Med. 69 (2018) 349–364.
[28] D. Stern, S. Du Yan, S.F. Yan, A.M. Schmidt, Receptor for advanced glycation
Supplementary material related to this article can be found, in the endproducts: a multiligand receptor magnifying cell stress in diverse pathologic
online version, at doi:https://doi.org/10.1016/j.phrs.2020.104792. settings, Adv. Drug Deliv. Rev. 54 (12) (2002) 1615–1625.
[29] K. Sugaya, T. Fukagawa, K.-i. Matsumoto, K. Mita, E.-i. Takahashi, A. Ando,
H. Inoko, T. Ikemura, Three genes in the human MHC class III region near the
References junction with the class II: gene for receptor of advanced glycosylation end products,
PBX2 homeobox gene and a notch homolog, human counterpart of mouse mam-
mary tumor gene int-3, Genomics 23 (2) (1994) 408–419.
[1] M.E. McCauley, R.H. Baloh, Inflammation in ALS/FTD pathogenesis, Acta
[30] R. Kang, R. Chen, Q. Zhang, W. Hou, S. Wu, L. Cao, J. Huang, Y. Yu, X.-g. Fan,
Neuropathol. 137 (5) (2019) 715–730.
Z. Yan, HMGB1 in health and disease, Mol. Aspects Med. 40 (2014) 1–116.
[2] J. Sreedharan, I.P. Blair, V.B. Tripathi, X. Hu, C. Vance, B. Rogelj, S. Ackerley,
[31] J. Derk, M. MacLean, J. Juranek, A.M. Schmidt, The receptor for advanced glyca-
J.C. Durnall, K.L. Williams, E. Buratti, TDP-43 mutations in familial and sporadic
tion endproducts (RAGE) and mediation of inflammatory neurodegeneration, J.
amyotrophic lateral sclerosis, Science 319 (5870) (2008) 1668–1672.
Alzheimers Dis. Parkinsonism 8 (1) (2018).
[3] O. Hardiman, A. Al-Chalabi, A. Chio, E.M. Corr, G. Logroscino, W. Robberecht,
[32] Y.K. Chuah, R. Basir, H. Talib, T.H. Tie, N. Nordin, Receptor for advanced glycation
P.J. Shaw, Z. Simmons, L.H. Van Den Berg, Amyotrophic lateral sclerosis, Nat. Rev.
end products and its involvement in inflammatory diseases, Int. J. Inflam. 2013
Dis. Primers 3 (2017) 17071.
(2013).
[4] R.H. Brown, A. Al-Chalabi, Amyotrophic lateral sclerosis, N. Engl. J. Med. 377 (2)
[33] S. Du Yan, X. Chen, J. Fu, M. Chen, H. Zhu, A. Roher, T. Slattery, L. Zhao,
(2017) 162–172.
M. Nagashima, J. Morser, RAGE and amyloid-β peptide neurotoxicity in
[5] A.E. Renton, A. Chiò, B.J. Traynor, State of play in amyotrophic lateral sclerosis
Alzheimer’s disease, Nature 382 (6593) (1996) 685.
genetics, Nat. Neurosci. 17 (1) (2014) 17.
[34] J. Gao, J. Teng, H. Liu, X. Han, B. Chen, A. Xie, Association of RAGE gene poly-
[6] D.R. Beers, S.H. Appel, Immune dysregulation in amyotrophic lateral sclerosis:
morphisms with sporadic Parkinson’s disease in Chinese Han population, Neurosci.
mechanisms and emerging therapies, Lancet Neurol. 18 (2) (2019) 211–220.
Lett. 559 (2014) 158–162.
[7] V. Kumar, T. Kashav, M.I. Hassan, Amyotrophic Lateral Sclerosis: Current
[35] J.K. Juranek, G.K. Daffu, J. Wojtkiewicz, D. Lacomis, J. Kofler, A.M. Schmidt,
Therapeutic Perspectives, Pathology, Prevention and Therapeutics of
Receptor for advanced glycation end products and its inflammatory ligands are
Neurodegenerative Disease, Springer, 2019, pp. 207–224.
upregulated in amyotrophic lateral sclerosis, Front. Cell. Neurosci. 9 (2015) 485.
[8] S.E. Parker, A.M. Hanton, S.N. Stefanou, P.G. Noakes, T.M. Woodruff, J.D. Lee,
[36] S. Akira, S. Uematsu, O. Takeuchi, Pathogen recognition and innate immunity, Cell
Revisiting the role of the innate immune complement system in ALS, Neurobiol. Dis.
124 (4) (2006) 783–801.
(2019).
[37] M. Boozari, A.E. Butler, A. Sahebkar, Impact of curcumin on toll‐like receptors, J.
[9] Y.N. Paudel, M. Shaikh, A. Chakraborti, Y. Kumari, Á. Aledo-Serrano,
Cell. Physiol. (2019).
K. Aleksovska, M.K.M. Alvim, I. Othman, HMGB1: a common biomarker and po-
[38] F. Peri, M. Piazza, Therapeutic targeting of innate immunity with Toll-like receptor
tential target for TBI, neuroinflammation, epilepsy, and cognitive dysfunction,
4 (TLR4) antagonists, Biotechnol. Adv. 30 (1) (2012) 251–260.
Front. Neurosci. 12 (2018) 628.
[39] H. Yang, H. Wang, Z. Ju, A.A. Ragab, P. Lundbäck, W. Long, S.I. Valdes-Ferrer,
[10] H.-M. Gao, H. Zhou, F. Zhang, B.C. Wilson, W. Kam, J.-S. Hong, HMGB1 acts on
M. He, J.P. Pribis, J. Li, MD-2 is required for disulfide HMGB1–dependent TLR4
microglia Mac1 to mediate chronic neuroinflammation that drives progressive
signaling, J. Exp. Med. (2015) jem. 20141318.
neurodegeneration, J. Neurosci. 31 (3) (2011) 1081–1092.
[40] H. Yang, H.S. Hreggvidsdottir, K. Palmblad, H. Wang, M. Ochani, J. Li, B. Lu,
[11] E. Angelopoulou, C. Piperi, A.G. Papavassiliou, High‐mobility group box 1 in
S. Chavan, M. Rosas-Ballina, Y. Al-Abed, A critical cysteine is required for HMGB1
Parkinson’s disease: from pathogenesis to therapeutic approaches, J. Neurochem.
binding to Toll-like receptor 4 and activation of macrophage cytokine release, Proc.
146 (3) (2018) 211–218.
Natl. Acad. Sci. 107 (26) (2010) 11942–11947.
[12] K. Takata, Y. Kitamura, D. Tsuchiya, T. Kawasaki, T. Taniguchi, S. Shimohama,
[41] U. Andersson, H. Yang, H. Harris, Extracellular HMGB1 as a therapeutic target in
High mobility group box protein‐1 inhibits microglial Aβ clearance and enhances
inflammatory diseases, Expert Opin. Ther. Targets 22 (3) (2018) 263–277.
Aβ neurotoxicity, J. Neurosci. Res. 78 (6) (2004) 880–891.
[42] W. Zhao, D.R. Beers, J.S. Henkel, W. Zhang, M. Urushitani, J.P. Julien, S.H. Appel,
[13] M. Maroso, S. Balosso, T. Ravizza, J. Liu, E. Aronica, A.M. Iyer, C. Rossetti,
Extracellular mutant SOD1 induces microglial‐mediated motoneuron injury, Glia 58
M. Molteni, M. Casalgrandi, A.A. Manfredi, Toll-like receptor 4 and high-mobility
(2) (2010) 231–243.
group box-1 are involved in ictogenesis and can be targeted to reduce seizures, Nat.
[43] U. Andersson, K.J. Tracey, HMGB1 is a therapeutic target for sterile inflammation
Med. 16 (4) (2010) 413.
and infection, Annu. Rev. Immunol. 29 (2011) 139–162.
[14] Y.N. Paudel, B.D. Semple, N.C. Jones, I. Othman, M.F. Shaikh, High mobility group
[44] K. Stark, V. Philippi, S. Stockhausen, J. Busse, A. Antonelli, M. Miller, I. Schubert,
box 1 (HMGB 1) as a novel frontier in epileptogenesis: from pathogenesis to ther-
P. Hoseinpour, S. Chandraratne, M.-L. von Brühl, Disulfide HMGB1 derived from
apeutic approaches, J. Neurochem. (2019).
platelets coordinates venous thrombosis in mice, Blood (2016) blood-2016-04-
[15] Å. Andersson, R. Covacu, D. Sunnemark, A.I. Danilov, A. Dal Bianco, M. Khademi,
710632.
E. Wallström, A. Lobell, L. Brundin, H. Lassmann, Pivotal advance: HMGB1 ex-
[45] H.J. Huttunen, C. Fages, J. Kuja-Panula, A.J. Ridley, H. Rauvala, Receptor for ad-
pression in active lesions of human and experimental multiple sclerosis, J. Leukoc.
vanced glycation end products-binding COOH-terminal motif of amphoterin in-
Biol. 84 (5) (2008) 1248–1255.
hibits invasive migration and metastasis, Cancer Res. 62 (16) (2002) 4805–4811.
[16] Y.N. Paudel, E. Angelopoulou, C. Piperi, I. Othman, High mobility group box 1
[46] N. D’Ambrosi, M. Cozzolino, M.T. Carri, Neuroinflammation in amyotrophic lateral
(HMGB1) protein in multiple sclerosis (MS): mechanisms and therapeutic potential,
sclerosis: role of redox (dys) regulation, Antioxid. Redox Signal. 29 (1) (2018)
Life Sci. (2019) 116924.
15–36.
[17] L. Brambilla, F. Martorana, G. Guidotti, D. Rossi, Dysregulation of astrocytic
[47] A. Motataianu, L. Barcutean, R. Balasa, Neuroimmunity in amyotrophic lateral
HMGB1 signaling in amyotrophic lateral sclerosis, Front. Neurosci. 12 (2018) 622.
sclerosis: focus on microglia, Amyotrophic Lateral Sclerosis and Frontotemporal
[18] J.D. Lee, N. Liu, S.C. Levin, L. Ottosson, U. Andersson, H.E. Harris, T.M. Woodruff,
Degeneration, (2020), pp. 1–8.
Therapeutic blockade of HMGB1 reduces early motor deficits, but not survival in
[48] D.L. Coco, P. Veglianese, E. Allievi, C. Bendotti, Distribution and cellular

9
Y.N. Paudel, et al. Pharmacological Research 156 (2020) 104792

localization of high mobility group box protein 1 (HMGB1) in the spinal cord of a inflammatory responses via TLR4 in brain glia, Am. J. Pathol. 168 (5) (2006)
transgenic mouse model of ALS, Neurosci. Lett. 412 (1) (2007) 73–77. 1619–1630.
[49] J.Y. Lee, J.D. Lee, S. Phipps, P.G. Noakes, T.M. Woodruff, Absence of toll-like re- [74] P. Xiang, T. Chen, Y. Mou, H. Wu, P. Xie, G. Lu, X. Gong, Q. Hu, Y. Zhang, H. Ji, NZ
ceptor 4 (TLR4) extends survival in the hSOD1 G93A mouse model of amyotrophic suppresses TLR4/NF-κB signalings and NLRP3 inflammasome activation in LPS-
lateral sclerosis, J. Neuroinflammation 12 (1) (2015) 90. induced RAW264. 7 macrophages, Inflamm. Res. 64 (10) (2015) 799–808.
[50] K. Yamanaka, S.J. Chun, S. Boillee, N. Fujimori-Tonou, H. Yamashita, [75] S.G. Crisafulli, S. Brajkovic, M.S.C. Mis, V. Parente, S. Corti, Therapeutic strategies
D.H. Gutmann, R. Takahashi, H. Misawa, D.W. Cleveland, Astrocytes as determi- under development targeting inflammatory mechanisms in amyotrophic lateral
nants of disease progression in inherited amyotrophic lateral sclerosis, Nat. sclerosis, Mol. Neurobiol. 55 (4) (2018) 2789–2813.
Neurosci. 11 (3) (2008) 251. [76] B. Parajuli, Y. Sonobe, J. Kawanokuchi, Y. Doi, M. Noda, H. Takeuchi, T. Mizuno,
[51] M. Nagai, D.B. Re, T. Nagata, A. Chalazonitis, T.M. Jessell, H. Wichterle, A. Suzumura, GM-CSF increases LPS-induced production of proinflammatory
S. Przedborski, Astrocytes expressing ALS-linked mutated SOD1 release factors se- mediators via upregulation of TLR4 and CD14 in murine microglia, J.
lectively toxic to motor neurons, Nat. Neurosci. 10 (5) (2007) 615. Neuroinflammation 9 (1) (2012) 268.
[52] F.P. Di Giorgio, M.A. Carrasco, M.C. Siao, T. Maniatis, K. Eggan, Non–cell auton- [77] A. Gugliandolo, S. Giacoppo, P. Bramanti, E. Mazzon, NLRP3 inflammasome acti-
omous effect of glia on motor neurons in an embryonic stem cell–based ALS model, vation in a transgenic amyotrophic lateral sclerosis model, Inflammation 41 (1)
Nat. Neurosci. 10 (5) (2007) 608. (2018) 93–103.
[53] F. Martorana, L. Brambilla, C.F. Valori, C. Bergamaschi, C. Roncoroni, E. Aronica, [78] M.S. Lee, Y.-J. Kim, Signaling pathways downstream of pattern-recognition re-
A. Volterra, P. Bezzi, D. Rossi, The BH4 domain of Bcl-XL rescues astrocyte de- ceptors and their cross talk, Annu. Rev. Biochem. 76 (2007) 447–480.
generation in amyotrophic lateral sclerosis by modulating intracellular calcium [79] J.D. Lee, J.Y. Lee, S.M. Taylor, P.G. Noakes, T.M. Woodruff, Innate immunity in
signals, Hum. Mol. Genet. 21 (4) (2011) 826–840. ALS, Amyotrophic Lateral Sclerosis, IntechOpen, 2012.
[54] P. Van Damme, E. Bogaert, M. Dewil, N. Hersmus, D. Kiraly, W. Scheveneels, [80] P.-O. Freskgård, E. Urich, Antibody therapies in CNS diseases, Neuropharmacology
I. Bockx, D. Braeken, N. Verpoorten, K. Verhoeven, Astrocytes regulate GluR2 ex- 120 (2017) 38–55.
pression in motor neurons and their vulnerability to excitotoxicity, Proc. Natl. Acad. [81] A.M. Schmidt, Soluble RAGEs—prospects for treating & tracking metabolic and
Sci. 104 (37) (2007) 14825–14830. inflammatory disease, Vascul. Pharmacol. 72 (2015) 1–8.
[55] M. Meyer, M.S. Kruse, L. Garay, A. Lima, P. Roig, H. Hunt, J. Belanoff, E.R. de Kloet, [82] M. De Paola, S.E. Sestito, A. Mariani, C. Memo, R. Fanelli, M. Freschi, C. Bendotti,
M.C.G. Deniselle, A.F. De Nicola, Long-term effects of the glucocorticoid receptor V. Calabrese, F. Peri, Synthetic and natural small molecule TLR4 antagonists inhibit
modulator CORT113176 in murine motoneuron degeneration, Brain Res. 1727 motoneuron death in cultures from ALS mouse model, Pharmacol. Res. 103 (2016)
(2020) 146551. 180–187.
[56] L. Duchen, S.J. Strich, An hereditary motor neurone disease with progressive de- [83] A. Fellner, Y. Barhum, A. Angel, N. Perets, I. Steiner, D. Offen, N. Lev, Toll-like
nervation of muscle in the mouse: the mutant’wobbler’, J. Neurol. Neurosurg. receptor-4 inhibitor TAK-242 attenuates motor dysfunction and spinal cord pa-
Psychiatr. 31 (6) (1968) 535. thology in an amyotrophic lateral sclerosis mouse model, Int. J. Mol. Sci. 18 (8)
[57] L.I. Bruijn, T.M. Miller, D.W. Cleveland, Unraveling the mechanisms involved in (2017) 1666.
motor neuron degeneration in ALS, Annu. Rev. Neurosci. 27 (2004) 723–749. [84] M. Dadon-Nachum, E. Melamed, D. Offen, The “dying-back” phenomenon of motor
[58] J.M. Moser, P. Bigini, T. Schmitt-John, The wobbler mouse, an ALS animal model, neurons in ALS, J. Mol. Neurosci. 43 (3) (2011) 470–477.
Mol. Genet. Genom. 288 (5-6) (2013) 207–229. [85] M. De Paola, A. Mariani, P. Bigini, M. Peviani, G. Ferrara, M. Molteni, S. Gemma,
[59] C.-S. Hwang, G.-T. Liu, M.D.-T. Chang, I.-L. Liao, H.-T. Chang, Elevated serum P. Veglianese, V. Castellaneta, V. Boldrin, Neuroprotective effects of toll-like re-
autoantibody against high mobility group box 1 as a potent surrogate biomarker for ceptor 4 antagonism in spinal cord cultures and in a mouse model of motor neuron
amyotrophic lateral sclerosis, Neurobiol. Dis. 58 (2013) 13–18. degeneration, Mol. Med. 18 (6) (2012) 971.
[60] A. Saleh, D.R. Smith, L. Tessler, A.R. Mateo, C. Martens, E. Schartner, R. Van der [86] A. Al-Chalabi, F. Fang, M.F. Hanby, P.N. Leigh, C.E. Shaw, W. Ye, F. Rijsdijk, An
Ploeg, C. Toth, D.W. Zochodne, P. Fernyhough, Receptor for advanced glycation estimate of amyotrophic lateral sclerosis heritability using twin data, Journal of
end-products (RAGE) activates divergent signaling pathways to augment neurite Neurology, Neurosurgery & Psychiatry 81 (12) (2010) 1324–1326.
outgrowth of adult sensory neurons, Exp. Neurol. 249 (2013) 149–159. [87] R.G. Miller, J.D. Mitchell, D.H. Moore, Riluzole for amyotrophic lateral sclerosis
[61] J. Juranek, R. Ray, M. Banach, V. Rai, Receptor for advanced glycation end-pro- (ALS)/motor neuron disease (MND), Cochrane Database Syst. Rev. (3) (2012).
ducts in neurodegenerative diseases, Rev. Neurosci. 26 (6) (2015) 691–698. [88] M.E. Gurney, T.J. Fleck, C.S. Himes, E.D. Hall, Riluzole preserves motor function in
[62] R. Ray, J.K. Juranek, V. Rai, RAGE axis in neuroinflammation, neurodegeneration a transgenic model of familial amyotrophic lateral sclerosis, Neurology 50 (1)
and its emerging role in the pathogenesis of amyotrophic lateral sclerosis, Neurosci. (1998) 62–66.
Biobehav. Rev. 62 (2016) 48–55. [89] J. Li, M. Sung, S.B. Rutkove, Electrophysiologic biomarkers for assessing disease
[63] M. MacLean, J. Derk, H.H. Ruiz, J.K. Juranek, R. Ramasamy, A.M. Schmidt, The progression and the effect of riluzole in SOD1 G93A ALS mice, PLoS One 8 (6)
Receptor for Advanced Glycation End Products (RAGE) and DIAPH1: implications (2013) e65976.
for vascular and neuroinflammatory dysfunction in disorders of the central nervous [90] F. De Giorgio, C. Maduro, E.M. Fisher, A. Acevedo-Arozena, Transgenic and phy-
system, Neurochem. Int. (2019). siological mouse models give insights into different aspects of amyotrophic lateral
[64] J.K. Juranek, G.K. Daffu, M.S. Geddis, H. Li, R. Rosario, B.J. Kaplan, L. Kelly, sclerosis, Dis. Model. Mech. 12 (1) (2019) dmm037424.
A.M. Schmidt, Soluble RAGE treatment delays progression of amyotrophic lateral [91] Z. Osking, J.I. Ayers, R. Hildebrandt, K. Skruber, H. Brown, D. Ryu, A.R. Eukovich,
sclerosis in SOD1 mice, Front. Cell. Neurosci. 10 (2016) 117. T.E. Golde, D.R. Borchelt, T.-A. Read, ALS-linked SOD1 mutants enhance neurite
[65] M.J. Kim, M.R. Vargas, B.A. Harlan, K.M. Killoy, L.E. Ball, S. Comte-Walters, outgrowth and branching in adult motor neurons, iScience 11 (2019) 294–304.
M. Gooz, Y. Yamamoto, J.S. Beckman, L. Barbeito, Nitration and glycation turn [92] P. Koshal, M. Kumar, S. Jamwal, P.K. Bansal, Animal Model of Amyotrophic Lateral
mature NGF into a toxic factor for motor neurons: a role for p75NTR and RAGE Sclerosis, Animal Models of Neurological Disorders, Springer, 2017, pp. 277–289.
signaling in ALS, Antioxid. Redox Signal. 28 (18) (2018) 1587–1602. [93] L. Fu, K. Liu, H. Wake, K. Teshigawara, T. Yoshino, H. Takahashi, S. Mori,
[66] S. Pinto, C. Cunha, M. Barbosa, A.R. Vaz, D. Brites, Exosomes from NSC-34 cells M. Nishibori, Therapeutic effects of anti-HMGB1 monoclonal antibody on pilo-
transfected with hSOD1-G93A are enriched in miR-124 and drive alterations in carpine-induced status epilepticus in mice, Sci. Rep. 7 (1) (2017) 1179.
microglia phenotype, Front. Neurosci. 11 (2017) 273. [94] A.P. Robinson, M.W. Caldis, C.T. Harp, G.E. Goings, S.D. Miller, High-mobility
[67] A. Serrano, C. Donno, S. Giannetti, M. Perić, P. Andjus, N. D’Ambrosi, F. Michetti, group box 1 protein (HMGB1) neutralization ameliorates experimental autoimmune
The astrocytic S100B protein with its receptor RAGE is aberrantly expressed in encephalomyelitis, J. Autoimmun. 43 (2013) 32–43.
SOD1G93A models, and its inhibition decreases the expression of proinflammatory [95] Y. Okuma, K. Liu, H. Wake, J. Zhang, T. Maruo, I. Date, T. Yoshino, A. Ohtsuka,
genes, Mediators Inflamm. 2017 (2017). N. Otani, S. Tomura, Anti–high mobility group box‐1 antibody therapy for trau-
[68] J. Iłżecka, Serum‐soluble receptor for advanced glycation end product levels in matic brain injury, Ann. Neurol. 72 (3) (2012) 373–384.
patients with amyotrophic lateral sclerosis, Acta Neurol. Scand. 120 (2) (2009) [96] Y. Okuma, H. Wake, K. Teshigawara, Y. Takahashi, T. Hishikawa, T. Yasuhara,
119–122. S. Mori, H.K. Takahashi, I. Date, M. Nishibori, Anti–High mobility group Box 1
[69] S. Süssmuth, A. Sperfeld, A. Hinz, J. Brettschneider, S. Endruhn, A. Ludolph, antibody therapy may prevent cognitive dysfunction after traumatic brain injury,
H. Tumani, CSF glial markers correlate with survival in amyotrophic lateral World Neurosurg. 122 (2019) e864–e871.
sclerosis, Neurology 74 (12) (2010) 982–987. [97] Yj. Li, L. Wang, B. Zhang, F. Gao, C.M. Yang, Glycyrrhizin, an HMGB1 inhibitor,
[70] K. Shobha, P. Alladi, A. Nalini, T. Sathyaprabha, T. Raju, Exposure to CSF from exhibits neuroprotective effects in rats after lithium‐pilocarpine‐induced status
sporadic amyotrophic lateral sclerosis patients induces morphological transforma- epilepticus, J. Pharm. Pharmacol. 71 (3) (2019) 390–399.
tion of astroglia and enhances GFAP and S100β expression, Neurosci. Lett. 473 (1) [98] A. Raucci, S. Di Maggio, F. Scavello, A. D’Ambrosio, M.E. Bianchi, M.C. Capogrossi,
(2010) 56–61. The Janus face of HMGB1 in heart disease: a necessary update, Cell. Mol. Life Sci.
[71] R. Bowser, M.R. Turner, J. Shefner, Biomarkers in amyotrophic lateral sclerosis: 76 (2) (2019) 211–229.
opportunities and limitations, Nat. Rev. Neurol. 7 (11) (2011) 631. [99] M. Nishibori, S. Mori, H.K. Takahashi, Anti-HMGB1 monoclonal antibody therapy
[72] S.-C. Tang, T.V. Arumugam, X. Xu, A. Cheng, M.R. Mughal, D.G. Jo, J.D. Lathia, for a wide range of CNS and PNS diseases, J. Pharmacol. Sci. (2019).
D.A. Siler, S. Chigurupati, X. Ouyang, Pivotal role for neuronal Toll-like receptors in [100] S. VanPatten, Y. Al-Abed, High mobility group box-1 (HMGb1): current wisdom
ischemic brain injury and functional deficits, Proc. Natl. Acad. Sci. 104 (34) (2007) and advancement as a potential drug target: miniperspective, J. Med. Chem. 61
13798–13803. (12) (2017) 5093–5107.
[73] I. Jou, J.H. Lee, S.Y. Park, H.J. Yoon, E.-H. Joe, E.J. Park, Gangliosides trigger

10

You might also like